Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 811
Filtrar
1.
J Biol Chem ; 300(4): 107127, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38432633

RESUMO

Regulators of G protein signaling (RGS) proteins constrain G protein-coupled receptor (GPCR)-mediated and other responses throughout the body primarily, but not exclusively, through their GTPase-activating protein activity. Asthma is a highly prevalent condition characterized by airway hyper-responsiveness (AHR) to environmental stimuli resulting in part from amplified GPCR-mediated airway smooth muscle contraction. Rgs2 or Rgs5 gene deletion in mice enhances AHR and airway smooth muscle contraction, whereas RGS4 KO mice unexpectedly have decreased AHR because of increased production of the bronchodilator prostaglandin E2 (PGE2) by lung epithelial cells. Here, we found that knockin mice harboring Rgs4 alleles encoding a point mutation (N128A) that sharply curtails RGS4 GTPase-activating protein activity had increased AHR, reduced airway PGE2 levels, and augmented GPCR-induced bronchoconstriction compared with either RGS4 KO mice or WT controls. RGS4 interacted with the p85α subunit of PI3K and inhibited PI3K-dependent PGE2 secretion elicited by transforming growth factor beta in airway epithelial cells. Together, these findings suggest that RGS4 affects asthma severity in part by regulating the airway inflammatory milieu in a G protein-independent manner.


Assuntos
Asma , Proteínas RGS , Animais , Humanos , Camundongos , Asma/metabolismo , Asma/genética , Asma/patologia , Broncoconstrição/genética , Dinoprostona/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Hipersensibilidade Respiratória/metabolismo , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/patologia , Proteínas RGS/metabolismo , Proteínas RGS/genética , Linhagem Celular
3.
Cell Mol Biol Lett ; 28(1): 102, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38066447

RESUMO

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a disease with high mortality and morbidity. Regulator of G protein signaling protein 6 (RGS6), identified as a tumor suppressor gene, has received increasing attention owing to its close relationship with oxidative stress and inflammation. However, the association between ARDS and RGS6 has not been reported. METHODS: Congruously regulated G protein-coupled receptor (GPCR)-related genes and differentially expressed genes (DEGs) in an acute lung injury (ALI) model were identified, and functional enrichment analysis was conducted. In an in vivo study, the effects of RGS6 knockout were studied in a mouse model of ALI induced by lipopolysaccharide (LPS). HE staining, ELISA, and immunohistochemistry were used to evaluate pathological changes and the degree of inflammation. In vitro, qRT‒PCR, immunofluorescence staining, and western blotting were used to determine the dynamic changes in RGS6 expression in cells. The RGS6 overexpression plasmid was constructed for transfection. qRT‒PCR was used to assess proinflammatory factors transcription. Western blotting and flow cytometry were used to evaluate apoptosis and reactive oxygen species (ROS) production. Organoid culture was used to assess the stemness and self-renewal capacity of alveolar epithelial type II cells (AEC2s). RESULTS: A total of 110 congruously regulated genes (61 congruously upregulated and 49 congruously downregulated genes) were identified among GPCR-related genes and DEGs in the ALI model. RGS6 was downregulated in vivo and in vitro in the ALI model. RGS6 was expressed in the cytoplasm and accumulated in the nucleus after LPS stimulation. Compared with the control group, we found higher mortality, more pronounced body weight changes, more serious pulmonary edema and pathological damage, and more neutrophil infiltration in the RGS6 knockout group upon LPS stimulation in vivo. Moreover, AEC2s loss was significantly increased upon RGS6 knockout. Organoid culture assays showed slower alveolar organoid formation, fewer alveolar organoids, and impaired development of new structures after passaging upon RGS6 knockout. In addition, RGS6 overexpression decreased ROS production as well as proinflammatory factor transcription in macrophages and decreased apoptosis in epithelial cells. CONCLUSIONS: RGS6 plays a protective role in ALI not only in early inflammatory responses but also in endogenous lung stem cell regeneration.


Assuntos
Lesão Pulmonar Aguda , Proteínas RGS , Síndrome do Desconforto Respiratório , Animais , Camundongos , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , Proteínas de Ligação ao GTP/efeitos adversos , Proteínas de Ligação ao GTP/metabolismo , Inflamação/patologia , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Síndrome do Desconforto Respiratório/genética , Síndrome do Desconforto Respiratório/metabolismo , Proteínas RGS/metabolismo
4.
Biol Direct ; 18(1): 78, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37986113

RESUMO

BACKGROUND: Regulator of G protein signaling 5 (RGS5), as a negative regulator of G protein-coupled receptor (GPCR) signaling, is highly expressed in arterial VSMCs and pericytes, which is involved in VSMC phenotypic heterogeneity and vascular remodeling in tumors. However, its role in normal and tumor vascular remodeling is controversial. METHODS: RGS5 knockout (Rgs5-KO) mice and RGS5 overexpression or knockdown in VSMCs in vivo by adeno-associated virus type 9 (AAV) carrying RGS5 cDNA or small hairpin RNA (shRNA) targeting RGS5 were used to determine the functional significance of RGS5 in vascular inflammation. RGS5 expression in the triple-negative (TNBCs) and non-triple-negative breast cancers (Non-TNBCs) was determined by immunofluorescent and immunohistochemical staining. The effect of breast cancer cell-conditioned media (BC-CM) on the pro-inflammatory phenotype of VSMCs was measured by phagocytic activity assays, adhesion assay and Western blot. RESULTS: We identified that knockout and VSMC-specific knockdown of RGS5 exacerbated accumulation and pyroptosis of pro-inflammatory VSMCs, resulting in vascular remodeling, which was negated by VSMC-specific RGS5 overexpression. In contrast, in the context of breast cancer tissues, the role of RGS5 was completely disrupted. RGS5 expression was increased in the triple-negative breast cancer (TNBC) tissues and in the tumor blood vessels, accompanied with an extensive vascular network. VSMCs treated with BC-CM displayed enhanced pro-inflammatory phenotype and higher adherent with macrophages. Furthermore, tumor-derived RGS5 could be transferred into VSMCs. CONCLUSIONS: These findings suggest that tumor microenvironment shifts the function of RGS5 from anti-inflammation to pro-inflammation and induces the pro-inflammatory phenotype of VSMCs that is favorable for tumor metastasis.


Assuntos
Neoplasias , Proteínas RGS , Camundongos , Animais , Proteínas RGS/genética , Proteínas RGS/metabolismo , Remodelação Vascular/genética , Músculo Liso Vascular/metabolismo , Microambiente Tumoral , Camundongos Knockout , Homeostase , Inflamação , Proliferação de Células
5.
Cell Commun Signal ; 21(1): 316, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37924113

RESUMO

G protein-coupled receptors (GPCRs) play a key role in regulating the homeostasis of the internal environment and are closely associated with tumour progression as major mediators of cellular signalling. As a diverse and multifunctional group of proteins, the G protein signalling regulator (RGS) family was proven to be involved in the cellular transduction of GPCRs. Growing evidence has revealed dysregulation of RGS proteins as a common phenomenon and highlighted the key roles of these proteins in human cancers. Furthermore, their differential expression may be a potential biomarker for tumour diagnosis, treatment and prognosis. Most importantly, there are few systematic reviews on the functional/mechanistic characteristics and clinical application of RGS family members at present. In this review, we focus on the G-protein signalling regulator (RGS) family, which includes more than 20 family members. We analysed the classification, basic structure, and major functions of the RGS family members. Moreover, we summarize the expression changes of each RGS family member in various human cancers and their important roles in regulating cancer cell proliferation, stem cell maintenance, tumorigenesis and cancer metastasis. On this basis, we outline the molecular signalling pathways in which some RGS family members are involved in tumour progression. Finally, their potential application in the precise diagnosis, prognosis and treatment of different types of cancers and the main possible problems for clinical application at present are discussed. Our review provides a comprehensive understanding of the role and potential mechanisms of RGS in regulating tumour progression. Video Abstract.


Assuntos
Neoplasias , Proteínas RGS , Humanos , Transdução de Sinais , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/metabolismo , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
6.
J Immunol ; 211(11): 1656-1668, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37850953

RESUMO

Regulatory T cells (Tregs) are critical mediators of immune tolerance and play a diametric role in cancer and autoimmunity. Tumor-infiltrating Tregs are often associated with poor prognosis in solid tumors because their enrichment in the tumor microenvironment contributes to immunosuppression. Conversely, dysregulation in the Treg compartment can disrupt self-tolerance, leading to autoimmunity. In the present study, we describe what is, to our knowledge, a novel regulator of Tregs, the GTPase activator regulator of G protein 1 (RGS1), demonstrating that RGS1-deficient human Tregs show downregulation of Treg-associated genes and are less immunosuppressive. These RGS1-deficient Tregs exhibit perturbations to the FOXP3-c-MYC transcriptional axis and downstream metabolic and autophagy programs by shifting their energy demands toward glycolysis and rendering them less autophagic. Taken together, RGS1 may serve as an apical node of Treg function by regulating the FOXP3-c-MYC transcriptional axis, thereby providing a therapeutic rationale for targeting RGS1 for treatment of cancer and autoimmune diseases.


Assuntos
Doenças Autoimunes , Neoplasias , Proteínas RGS , Humanos , Linfócitos T Reguladores , Doenças Autoimunes/patologia , Autoimunidade , Neoplasias/patologia , Autofagia/genética , Fatores de Transcrição Forkhead/metabolismo , Microambiente Tumoral , Proteínas RGS/genética , Proteínas RGS/metabolismo
7.
Oncoimmunology ; 12(1): 2260620, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37781234

RESUMO

Colorectal cancer (CRC) remains a leading cause of cancer-related mortality despite efforts to improve standard interventions. As CRC patients can benefit from immunotherapeutic strategies that incite effector T cell action, cancer vaccines represent a safe and promising therapeutic approach to elicit protective and durable immune responses against components of the tumor microenvironment (TME). In this study, we investigate the pre-clinical potential of a Listeria monocytogenes (Lm)-based vaccine targeting the CRC-associated vasculature. CRC survival and progression are reliant on functioning blood vessels to effectively mediate various metabolic processes and oxygenate underlying tissues. We, therefore, advance the strategy of initiating immunity in syngeneic mouse models against the endogenous pericyte antigen RGS5, which is a critical mediator of pathological vascularization. Overall, Lm-based vaccination safely induced potent anti-tumor effects that consisted of recruiting functional Type-1-associated T cells into the TME and reducing tumor blood vessel content. This study underscores the promising clinical potential of targeting RGS5 against vascularized tumors like CRC.


Assuntos
Neoplasias do Colo , Listeria monocytogenes , Listeria , Proteínas RGS , Camundongos , Animais , Humanos , Pericitos , Neoplasias do Colo/prevenção & controle , Listeria monocytogenes/metabolismo , Vacinação , Microambiente Tumoral , Proteínas RGS/genética , Proteínas RGS/metabolismo
8.
Clin Exp Hypertens ; 45(1): 2271186, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37879890

RESUMO

Objective: Excessive proliferation and migration of pulmonary arterial smooth muscle cell (PASMC) is a core event of pulmonary hypertension (PH). Regulators of G protein signaling 10 (RGS10) can regulate cellular proliferation and cardiopulmonary diseases. We demonstrate whether RGS10 also serves as a regulator of PH.Methods: PASMC was challenged by hypoxia to induce proliferation and migration. Adenovirus carrying Rgs10 gene (Ad-Rgs10) was used for external expression of Rgs10. Hypoxia/SU5416 or MCT was used to induce PH. Right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI) were used to validate the establishment of PH model.Results: RGS10 was downregulated in hypoxia-challenged PASMC. Ad-Rgs10 significantly suppressed proliferation and migration of PASMC after hypoxia stimulus, while silencing RGS10 showed contrary effect. Mechanistically, we observed that phosphorylation of S6 and 4E-Binding Protein 1 (4EBP1), the main downstream effectors of mammalian target of rapamycin complex 1 (mTORC1) as well as phosphorylation of AKT, the canonical upstream of mTORC1 in hypoxia-induced PASMC were negatively modulated by RGS10. Both recovering mTORC1 activity and restoring AKT activity abolished these effects of RGS10 on PASMC. More importantly, AKT activation also abolished the inhibitory role of RGS10 in mTORC1 activity in hypoxia-challenged PASMC. Finally, we also observed that overexpression of RGS10 in vivo ameliorated pulmonary vascular wall thickening and reducing RVSP and RVHI in mouse PH model.Conclusion: Our findings reveal the modulatory role of RGS10 in PASMC and PH via AKT/mTORC1 axis. Therefore, targeting RGS10 may serve as a novel potent method for the prevention against PH."


Assuntos
Hipertensão Pulmonar , Proteínas RGS , Animais , Camundongos , Proliferação de Células , Células Cultivadas , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/farmacologia , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita , Hipóxia/metabolismo , Mamíferos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/farmacologia , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar , Proteínas RGS/genética , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia
9.
Genes (Basel) ; 14(9)2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37761960

RESUMO

Cancer metastasis accounts for approximately 90% of cancer deaths, and elucidating markers in metastasis is the first step in its prevention. To characterize metastasis marker genes (MGs) of breast cancer, XGBoost models that classify metastasis status were trained with gene expression profiles from TCGA. Then, a metastasis score (MS) was assigned to each gene by calculating the inner product between the feature importance and the AUC performance of the models. As a result, 54, 202, and 357 genes with the highest MS were characterized as MGs by empirical p-value cutoffs of 0.001, 0.005, and 0.01, respectively. The three sets of MGs were compared with those from existing metastasis marker databases, which provided significant results in most comparisons (p-value < 0.05). They were also significantly enriched in biological processes associated with breast cancer metastasis. The three MGs, SPPL2C, KRT23, and RGS7, showed highly significant results (p-value < 0.01) in the survival analysis. The MGs that could not be identified by statistical analysis (e.g., GOLM1, ELAVL1, UBP1, and AZGP1), as well as the MGs with the highest MS (e.g., ZNF676, FAM163B, LDOC2, IRF1, and STK40), were verified via the literature. Additionally, we checked how close the MGs were to each other in the protein-protein interaction networks. We expect that the characterized markers will help understand and prevent breast cancer metastasis.


Assuntos
Neoplasias da Mama , Segunda Neoplasia Primária , Proteínas RGS , Humanos , Feminino , Neoplasias da Mama/patologia , Transcriptoma , Mapas de Interação de Proteínas , Aprendizado de Máquina , Proteínas de Membrana/genética , Proteínas RGS/genética , Melanoma Maligno Cutâneo
10.
J Neuroinflammation ; 20(1): 203, 2023 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-37674228

RESUMO

Astrocytes contribute to chronic neuroinflammation in a variety of neurodegenerative diseases, including Parkinson's disease (PD), the most common movement disorder. However, the precise role of astrocytes in neuroinflammation remains incompletely understood. Herein, we show that regulator of G-protein signaling 5 (RGS5) promotes neurodegenerative process through augmenting astrocytic tumor necrosis factor receptor (TNFR) signaling. We found that selective ablation of Rgs5 in astrocytes caused an inhibition in the production of cytokines resulting in mitigated neuroinflammatory response and neuronal survival in animal models of PD, whereas overexpression of Rgs5 had the opposite effects. Mechanistically, RGS5 switched astrocytes from neuroprotective to pro-inflammatory property via binding to the receptor TNFR2. RGS5 also augmented TNFR signaling-mediated pro-inflammatory response by interacting with the receptor TNFR1. Moreover, interrupting RGS5/TNFR interaction by either RGS5 aa 1-108 or small molecular compounds feshurin and butein, suppressed astrocytic cytokine production. We showed that the transcription of astrocytic RGS5 was controlled by transcription factor early B cell factor 1 whose expression was reciprocally influenced by RGS5-modulated TNF signaling. Thus, our study indicates that beyond its traditional role in G-protein coupled receptor signaling, astrocytic RGS5 is a key modulator of TNF signaling circuit with resultant activation of astrocytes thereby contributing to chronic neuroinflammation. Blockade of the astrocytic RGS5/TNFR interaction is a potential therapeutic strategy for neuroinflammation-associated neurodegenerative diseases.


Assuntos
Doenças Neuroinflamatórias , Proteínas RGS , Animais , Astrócitos , Transdução de Sinais , Proteínas RGS/genética , Inflamação
11.
Int J Oncol ; 63(4)2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37594134

RESUMO

Breast cancer (BRCA) exhibits a high incidence rate among women worldwide. LOC127814295 (ENSG00000232995), termed long non­coding (lnc)­regulator of G protein signaling 5 (RGS5), is a novel lncRNA with a genomic region overlapping with protein­coding gene RGS5. Results obtained using The Cancer Genome Atlas demonstrated that lnc­RGS5 was deregulated in diverse cancer types, including BRCA; however, the functional role of lnc­RGS5 remains unclear. Results of the present study demonstrated that lnc­RGS5 was upregulated in BRCA tissues compared with healthy samples (n=30; P<0.0001), and was associated with the overall survival of patients with triple­negative BRCA (n=106; P<0.05). Moreover, lnc­RGS5 expression was significantly higher in triple­negative BRCA samples than in LumA, LumB, or Her2 subtypes (P<0.05). Functionally, lnc­RGS5 upregulation promoted BRCA cell proliferation in vitro, whereas lnc­RGS5 knockdown elicited the opposite function. Stable knockdown of lnc­RGS5 inhibited tumor cell proliferation in vivo. Bioinformatics analysis revealed that lnc­RGS5 was significantly associated with RNA binding involved in post­transcriptional gene silencing (P=0.002). Mechanistically, lnc­RGS5 functions as a competing endogenous RNA via competitively sponging miR­542­5p to upregulate forkhead box M1 (FoxM1) and the VEGFA/Neuropilin 1 axis; thus, promoting BRCA cell proliferation in vitro. Moreover, rescue experiments validated that the lnc­RGS5/miR­542­5p/FoxM1 axis promoted BRCA cell growth in vivo. Collectively, results of the present study demonstrated that lnc­RGS5 may exhibit potential as a novel oncogenic lncRNA in BRCA. The present study may provide a novel theoretical basis for the role of lncRNA in the targeted therapy of BRCA.


Assuntos
MicroRNAs , Proteínas RGS , RNA Longo não Codificante , Neoplasias de Mama Triplo Negativas , Humanos , Feminino , RNA Longo não Codificante/genética , Transdução de Sinais/genética , Proliferação de Células/genética , MicroRNAs/genética , Fator A de Crescimento do Endotélio Vascular , Proteínas RGS/genética
12.
J Transl Med ; 21(1): 583, 2023 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-37649067

RESUMO

Currently, women around the world are still suffering from various female common diseases with the high incidence, such as ovarian cancer, uterine fibroids and preeclampsia (PE), and some diseases are even with the high mortality rate. As a negative feedback regulator in G Protein-Coupled Receptor signaling (GPCR), the Regulator of G-protein Signaling (RGS) protein family participates in regulating kinds of cell biological functions by destabilizing the enzyme-substrate complex through the transformation of hydrolysis of G Guanosine Triphosphate (GTP). Recent work has indicated that, the Regulator of G-protein Signaling 2 (RGS2), a member belonging to the RGS protein family, is closely associated with the occurrence and development of certain female diseases, providing with the evidence that RGS2 functions in sustaining women's health. In this review paper, we summarize the current knowledge of RGS2 in female common diseases, and also tap and discuss its therapeutic potential by targeting multiple mechanisms.


Assuntos
Leiomioma , Proteínas RGS , Gravidez , Humanos , Feminino , Saúde da Mulher , Transdução de Sinais , Hidrólise , Conhecimento
13.
FASEB J ; 37(8): e23064, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37440271

RESUMO

Off target damage to vital organ systems is an unfortunate side effect of cancer chemotherapy and remains a major limitation to the use of these essential drugs in the clinic. Despite decades of research, the mechanisms conferring susceptibility to chemotherapy driven cardiotoxicity and hepatotoxicity remain unclear. In the livers of patients with a history of chemotherapy, we observed a twofold increase in expression of G protein regulator RGS7 and a corresponding decrease in fellow R7 family member RGS11. Knockdown of RGS7 via introduction of RGS7 shRNA via tail vein injection decreased doxorubicin-induced hepatic collagen and lipid deposition, glycogen accumulation, and elevations in ALT, AST, and triglycerides by approximately 50%. Surprisingly, a similar result could be achieved via introduction of RGS7 shRNA directly to the myocardium without impacting RGS7 levels in the liver directly. Indeed, doxorubicin-treated cardiomyocytes secrete the endocrine factors transforming growth factor ß1 (TGFß1) and TGFß superfamily binding protein follistatin-related protein 1 (FSTL1). Importantly, RGS7 overexpression in the heart was sufficient to recapitulate the impacts of doxorubicin on the liver and inhibition of TGFß1 signaling with the receptor blocker GW788388 ameliorated the effect of cardiac RGS7 overexpression on hepatic fibrosis, steatosis, oxidative stress, and cell death as well as the resultant elevation in liver enzymes. Together these data demonstrate that RGS7 controls both the release of TGFß1 from the heart and the profibrotic and pro-oxidant actions of TGFß1 in the liver and emphasize the functional significance of endocrine cardiokine signaling in the pathogenesis of chemotherapy drive multiorgan damage.


Assuntos
Proteínas Relacionadas à Folistatina , Proteínas RGS , Humanos , Proteínas RGS/genética , Proteínas RGS/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Transporte/metabolismo , Fígado/metabolismo , Doxorrubicina/efeitos adversos , Proteínas Relacionadas à Folistatina/metabolismo
14.
Osteoporos Int ; 34(11): 1907-1916, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37500982

RESUMO

Bone mineral density (BMD) is an essential predictor of osteoporosis and fracture. We conducted a genome-wide trajectory analysis of BMD and analyzed the BMD change. PURPOSE: This study aimed to identify the genetic architecture and potential biomarkers of BMD. METHODS: Our analysis included 141,261 white participants from the UK Biobank with heel BMD phenotype data. We used a genome-wide trajectory analysis tool, TrajGWAS, to conduct a genome-wide association study (GWAS) of BMD. Then, we validated our findings in previously reported BMD genetic associations and performed replication analysis in the Asian participants. Finally, gene-set enrichment analysis (GSEA) of the identified candidate genes was conducted using the FUMA platform. RESULTS: A total of 52 genes associated with BMD trajectory mean were identified, of which the top three significant genes were WNT16 (P = 1.31 × 10-126), FAM3C (P = 4.18 × 10-108), and CPED1 (P = 8.48 × 10-106). In addition, 114 genes associated with BMD within-subject variability were also identified, such as AC092079.1 (P = 2.72 × 10-13) and RGS7 (P = 4.72 × 10-10). The associations for these candidate genes were confirmed in the previous GWASs and replicated successfully in the Asian participants. GSEA results of BMD change identified multiple GO terms related to skeletal development, such as SKELETAL SYSTEM DEVELOPMENT (Padjusted = 2.45 × 10-3) and REGULATION OF OSSIFICATION (Padjusted = 2.45 × 10-3). KEGG enrichment analysis showed that these genes were mainly enriched in WNT SIGNALING PATHWAY. CONCLUSIONS: Our findings indicated that the CPED1-WNT16-FAM3C locus plays a significant role in BMD mean trajectories and identified several novel candidate genes contributing to BMD within-subject variability, facilitating the understanding of the genetic architecture of BMD.


Assuntos
Osteoporose , Proteínas RGS , Humanos , Densidade Óssea/genética , Estudo de Associação Genômica Ampla , Bancos de Espécimes Biológicos , Osteoporose/genética , Reino Unido , Polimorfismo de Nucleotídeo Único , Proteínas RGS/genética , Proteínas de Neoplasias/genética , Citocinas
15.
PLoS One ; 18(7): e0288357, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37437037

RESUMO

INTRODUCTION: Prognostic factors in plasma cell myeloma were proved to be related to signaling pathways and associated transcription factors. RGS1 and mTOR were known to play an important role in the pathogenesis of multiple myeloma. The aim of the study was to evaluate the expression and the prognostic value of RGS1 and mTOR and their relation to clinical as well as other diagnostic criteria in multiple myeloma. PATIENTS AND METHODS: The present study included 44 denovo Myeloma patients, recruited from the Medical Oncology Department, National Cancer Institute, Cairo University. Detection of RGS1 and mTOR expression was performed using Immunohistochemical staining on bone marrow biopsy sections. RESULTS: The median age was 51 years with male to female ratio 1.58:1. There was a positive highly statistically significant correlation between RGS1 and mTOR among all studied cases (p value <0.001). Regarding their prognostic value, there was a highly statistically significant association of the expression levels of RGS1 and mTOR with treatment response (p <0.001). Finally, there was a significant influence of RGS1 and mTOR on overall survival probability (p value <0.001 and <0.002 respectively) with better survival for those having low expression. CONCLUSION: RGS1 and mTOR were suggested as poor prognostic markers in MM patients, being associated with lower response rate and inferior OS. We recommend considering RGS1 and mTOR as one of the prognostic criteria in different risk stratification and staging classifications. Further trials for RGS1 and mTOR targeting in multiple myeloma are recommended.


Assuntos
Mieloma Múltiplo , Proteínas RGS , Humanos , Feminino , Masculino , Pessoa de Meia-Idade , Prognóstico , Mieloma Múltiplo/diagnóstico , Egito , Serina-Treonina Quinases TOR , Biópsia , Proteínas RGS/genética
16.
Sci Rep ; 13(1): 9519, 2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-37308506

RESUMO

Venous thromboembolism (VTE) is a leading cause of death among cancer patients. Khorana score (KS) is the most studied tool to predict cancer-related VTE, however, it exerts poor sensitivity. Several single-nucleotide polymorphisms (SNPs) have been associated with VTE risk in the general population, but whether they are predictors of cancer-related VTE is a matter of discussion. Compared to other solid tumours, little is known about VTE in the setting of cervical cancer (CC) and whether thrombogenesis-related polymorphisms could be valuable biomarkers in patients with this neoplasia. This study aims to analyse the effect of VTE occurrence on the prognosis of CC patients, explore the predictive capability of KS and the impact of thrombogenesis-related polymorphisms on CC-related VTE incidence and patients' prognosis regardless of VTE. A profile of eight SNPs was evaluated. A retrospective hospital-based cohort study was conducted with 400 CC patients under chemoradiotherapy. SNP genotyping was carried on by using TaqMan® Allelic Discrimination methodology. Time to VTE occurrence and overall survival were the two measures of clinical outcome evaluated. The results indicated that VTE occurrence (8.5%) had a significant impact on the patient's survival (log-rank test, P < 0.001). KS showed poor performance (KS ≥ 3, χ2, P = 0.191). PROCR rs10747514 and RGS7 rs2502448 were significantly associated with the risk of CC-related VTE development (P = 0.021 and P = 0.006, respectively) and represented valuable prognostic biomarkers regardless of VTE (P = 0.004 and P = 0.010, respectively). Thus, thrombogenesis-related genetic polymorphisms may constitute valuable biomarkers among CC patients allowing a more personalized clinical intervention.


Assuntos
Proteínas RGS , Neoplasias do Colo do Útero , Tromboembolia Venosa , Humanos , Feminino , Estudos de Coortes , Estudos Retrospectivos , Prognóstico , Polimorfismo de Nucleotídeo Único
18.
Cell Signal ; 107: 110682, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37075876

RESUMO

Regulator of protein signaling (RGS20) is a member of the RGS protein superfamily, which serve as key negative regulators of G protein-mediated signal transduction. Through their GTPase accelerating protein (GAP) activity, RGS proteins deactivate α-subunits of heterotrimeric G proteins. In addition, the majority of RGS proteins also have the ability to act through other, non-GAP related, functions. RGS20 is one of three members of the RZ subfamily, which all show selective GAP activity towards Gαz, however emerging data suggest that RGS20 can also regulate Gi/o-mediated signaling. While increased RGS20 expression is associated with the progression of multiple cancers, a large gap still exists relating to the mechanisms of RGS20 regulation and function. RGS20 contains a poly-cysteine string motif and a conserved cysteine in RGS domain, which are assumed to be palmitoylated. Palmitoylation, an important post-translational modification, plays an important role in cells by changing cellular functions of proteins. Consequently, the aim of this study was to confirm that RGS20 is palmitoylated and determine how palmitoylation affects its inhibition of Gαo-mediated signaling. We found a significant positive correlation between RGS20 palmitoylation and its association with active Gαo. We also showed that a conserved cysteine residue in the RGS domain is a critical site for its palmitoylation, with large impact on its association with Gαo. Palmitoylation on this site did not affect its GAP activity, however, it increased the inhibition of Gαo-mediated cAMP signaling. Altogether these data suggest that palmitoylation is a regulatory mechanism controlling RGS20 function, and that RGS20 can inhibit Gαo signaling through both GAP activity and non-GAP mechanisms.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP , Proteínas RGS , Proteínas RGS/metabolismo , Lipoilação , Cisteína/metabolismo , Transdução de Sinais , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo
19.
Curr Mol Med ; 23(9): 981-990, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37073154

RESUMO

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease involving both cartilage and synovium. Activating transcription factor 3 (ATF3) and regulator of G protein signaling 1 (RGS1) have been reported to be up-regulated in OA. However, little is known regarding the relationship between these two genes and the mechanism of this relationship in OA development. Therefore, the present study explores the mechanism of ATF3-mediated RGS1 in the proliferation, migration, and apoptosis of synovial fibroblasts. METHODS: After the OA cell model was constructed with TGF-ß1 induction, human fibroblast-like synoviocytes (HFLSs) were transfected with ATF3 shRNA or RGS1 shRNA alone or co-transfected with ATF3 shRNA and pcDNA3.1-RGS1. Then, proliferation, migration, apoptosis, and the expression of ATF3, RGS1, α-SMA, BCL-2, caspase3, and cleaved-caspase3 were measured. Meanwhile, the potential relationship between ATF3 and RGS1 was predicted and validated. RESULTS AND DISCUSSION: Analysis of the GSE185059 dataset suggested that RGS1 was up-regulated in OA synovial fluid exosomes. Moreover, ATF3 and RGS1 were both highly expressed in TGF-ß1-induced HFLSs. Transfection of ATF3 shRNA or RGS1 shRNA significantly reduced proliferation and migration and promoted apoptosis of TGF- ß1-induced HFLSs. Mechanistically, ATF3 bound to the RGS1 promoter and elevated RGS1 expression. Silencing ATF3 repressed proliferation and migration and enhanced apoptosis of TGF-ß1-induced HFLSs by down-regulating RGS1. CONCLUSION: ATF3 binds to the RGS1 promoter and enhances RGS1 expression to accelerate cell proliferation and block cell apoptosis in TGF-ß1-induced synovial fibroblasts.


Assuntos
Proteínas RGS , Fator de Crescimento Transformador beta1 , Humanos , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Artroscopia , Fibroblastos/metabolismo , Apoptose/genética , Proliferação de Células/genética , RNA Interferente Pequeno/metabolismo , Células Cultivadas , Proteínas RGS/genética , Proteínas RGS/metabolismo
20.
Ann Clin Lab Sci ; 53(2): 238-247, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37094862

RESUMO

OBJECTIVE: Abdominal aortic aneurysm (AAA) is characterized by vascular smooth muscle cell (VSMC) injury. Circ_0000285 has been declared to drive cancer development, but its role in AAA remains unclear. We thus intended to disclose circ_0000285's role and molecular mechanism in AAA. METHODS: VSMCs were exposed to hydrogen peroxide (H2O2) to induce cell injury. Circ_0000285, miR-599, and regulator of G protein signaling 17 (RGS17) mRNA expressions were ascertained by conducting RT-qPCR assay while the levels of RGS17 protein was ascertained via western blotting. MiR-599's predicted binding with circ_0000285 and RGS17 were validated by means of the dual-luciferase reporter experiment. Cell proliferation was evaluated through the CCK-8 and EdU assays. Cell apoptosis was assessed via the caspase-3 activity assay. RESULTS: The AAA samples and H2O2-treated VSMCs manifested high expressions of circ_0000285 and RGS17 as well as a poor miR-599 expression. H2O2 treatment impaired the proliferation of VSMCs while stimulating their apoptosis. Circ_0000285 overexpression further repressed cell proliferation and enhanced apoptosis in H2O2-treated VSMCs while miR-599 enrichment partly reversed these effects. Circ_0000285 directly bound to miR-599, and miR-599 interacted with RGS17 3'UTR. RGS17 overexpression also suppressed cell proliferation and stimulated apoptosis in H2O2-treated VSMCs. Nevertheless, these effects were offset by miR-599 enrichment. CONCLUSION: Circ_0000285 governed the miR-599/RGS17 network to regulate H2O2-induced VSMC injuries, thereby promoting the development of AAA.


Assuntos
Aneurisma da Aorta Abdominal , MicroRNAs , Proteínas RGS , Humanos , MicroRNAs/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Peróxido de Hidrogênio/farmacologia , Transdução de Sinais , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Proliferação de Células/genética , Apoptose , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA